The upgraded role of HER3 and HER4 receptors in breast cancer

https://doi.org/10.1016/j.critrevonc.2009.04.011Get rights and content

Abstract

The human epidermal growth factor receptor (HER) family comprises four homologous members. The activation of these receptors affects essential tumorigenic processes and plays a crucial role in the pathogenesis of breast cancer. Among HER family members, EGFR and HER2 are the most studied. However, accumulating data provide evidence for the significance of HER3 and HER4 alterations in breast carcinogenesis. The combination of HER2 and HER3 receptors may be critical in breast cancer growth and progression. Moreover, HER3 may provide a route for resistance to agents targeting EGFR or HER2. Although a number of studies have demonstrated that HER3 overexpression is associated with poor prognosis in patients with breast cancer, other studies have indicated that HER3 overexpression may be a positive prognostic factor. With respect to HER4 receptor, the existing evidence suggests that HER4 signalling promotes differentiation and growth inhibition of breast cancer cells. In addition, HER4 is more consistently related with a favourable prognosis in breast cancer. HER4 has multiple different activities in the breast, and many of these functions are mediated by a soluble HER4 intracellular domain. In addition, loss of HER4 expression may represent a marker for resistance to tamoxifen. Because of the functional interdependency among the HER receptors, it is possible that the effect on cell proliferation and tumor growth depends on receptor trans-signalling. Therefore, clarifying how and the extent to which these different signalling pathways interact in breast carcinogenesis, may lead to additional therapeutic opportunities. This review presents an update on the role of HER3 and HER4 receptors in breast cancer. Moreover, we provide current data relating to the prognostic significance of these receptors, as well as their impact on the activity of HER-targeting therapies in patients with breast cancer.

Introduction

The human epidermal growth factor receptor (HER) family consists of four members: ErbB-1 [epidermal growth factor (EGF) receptor (EGFR) or HER1], ErbB-2 (HER2) for which no ligand has been described thus far, ErbB-3 (HER3) and ErbB-4 (HER4). The shared general structure of HER receptors consists of an extracellular ligand-binding domain (ECD), a transmembrane region, and an intracellular domain containing the tyrosine kinase and a carboxy-terminal region. The HER3 receptor is unique in the family in that it is catalytically inactive. Despite their structural homology, HER receptors differ in their ligand specificities. Two main ligand classes have to date been identified: the splice variants of neuregulins (NRGs) that bind exclusively to HER3 and/or HER4 and different EGF-related proteins [1]. Binding of specific ligands to domains I and III is followed by conformational changes of the ECD, exposing domain II, which facilitates receptor dimerization. Additional receptor interactions in extracellular, transmembrane and kinase domains, further stabilize the dimer. Dimerization results in the activation of the kinase domain, and the induction of intracellular signalling cascades that mediate cell growth and survival. Signalling diversity depends both on the presence of specific receptors and the characteristics of individual ligands. EGFR and HER2 classically couple to Ras-Raf-MEK-mitogen-activated protein kinase (MAPK)-dependent pathway, whereas HER3 is a potent activator of phosphatidylinositol 3-kinase (PI3-K)-Akt [2], [3]. In terms of activity, the HER family is characterized by a functional interdependency among its members.

Section snippets

Search strategy and selection criteria

References for this review were identified by searches in PubMed from 1990 until December 2008, including the terms “ErbB receptors and breast cancer”, “ErbB3 and breast cancer”, “HER3 and breast cancer”, “ErbB4 and breast cancer”, “HER4 and breast cancer”, “ErbB3 and breast” and “ErbB4 and breast”. Articles were also identified through searches of the authors’ own files. Only papers published in English were reviewed.

HER3 and breast cancer

The HER3 gene is located on chromosome 12q13 and the encoded protein receptor binds to NRG-1 and NRG-2. Overexpression of HER3 receptor has been reported in 20–30% of invasive breast carcinomas [4]. HER3 signalling relies on the formation of heterodimers with other members of the HER family, since HER3 receptor has no intrinsic kinase activity. Although it is kinase defective, HER3 can be phosphorylated by other receptors such as HER2. Neither ligandless HER2 nor tyrosine kinase deficient HER3

HER4 and breast cancer

The HER4 gene is located on chromosome 2q33.3–34 and the encoded protein can be activated by both NRGs and some ligands of the EGF family (betacellulin, epiregulin, heparin-binding EGF-like ligand). HER4 expression is detectable in less than half of breast cancers. In contrast to the other HER receptors, the existing evidence suggests that HER4 is characterized by antiproliferative and pro-apoptotic activity [31], [32]. In cell line experiments, when HER2 positive cancer cells were transfected

Evaluating the HER family as a whole

As we have mentioned elsewhere [47], the majority of clinicopathological studies have focused on the expression and/or gene amplification of individual HER family members. Because of the complex interactions among the HER receptors, it is possible that the effect on cell proliferation and tumor growth depends on receptor trans-signalling and hence, the evaluation of the combined expression pattern of all family members merits further scrutiny. Limited data are available on the expression

Conclusion

Studies that have examined the HER family, either at the protein- or the mRNA level, have demonstrated a complex expression profile of HER receptors in patients with breast cancer. Beyond any doubt the HER family constitutes an attractive field for the development of targeted therapies and significant treatment advances have been made thus far. However, the failure of HER-targeted strategies in a substantial proportion of patients with HER2-overexpressing breast tumors reinforces the need to

Reviewers

Prof. David A. Cameron, Cookridge Hospital, NCRN Coordinating Centre, Hospital Lane, Leeds LS16 6QB, United Kingdom.

Dr. Françoise Révillion, Centre Oscar Lambret, Laboratoire d’Oncologie Moléculaire Humaine, 3 rue Frédéric Combemale, F-59020 Lille Cedex, France.

Conflict of interest statement

None.

Angelos K. Koutras, M.D., Ph.D., is a consultant medical oncologist in the Division of Medical Oncology of the University Hospital of Patras, Greece. He is also a senior research fellow at the Laboratory of Clinical Oncology of the University Hospital of Patras, Greece. His main clinical interests include breast, colorectal and lung cancer. His research interests focus on human epidermal growth factor receptor signalling pathways in breast cancer, as well as hormonal signalling in non-small

References (49)

  • S.P. Soltoff et al.

    ErbB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor

    Mol Cell Biol

    (1994)
  • D.F. Stern

    ERBB3/HER3 and ERBB2/HER2 duet in mammary development and breast cancer

    J Mammary Gland Biol Neoplasia

    (2008)
  • T. Holbro et al.

    The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation

    Proc Natl Acad Sci USA

    (2003)
  • R. Naidu et al.

    Expression of c-erbB3 protein in primary breast carcinomas

    Br J Cancer

    (1998)
  • I. Bieche et al.

    Prognostic value of ERBB family mRNA expression in breast carcinomas

    Int J Cancer

    (2003)
  • C.J. Witton et al.

    Expression of the HER1-4 family of receptor tyrosine kinases in breast cancer

    J Pathol

    (2003)
  • A. Sassen et al.

    Cytogenetic analysis of HER1/EGFR, HER2, HER3, and HER4 in 278 breast cancer patients

    Breast Cancer Res

    (2008)
  • N.R. Lemoine et al.

    Expression of the ERBB3 gene product in breast cancer

    Br J Cancer

    (1992)
  • C.M. Quinn et al.

    C-erbB-3 protein expression in human breast cancer: comparison with other tumour variables and survival

    Histopathology

    (1994)
  • A. Travis et al.

    C-erbB-3 in human breast carcinoma: expression and relation to prognosis and established prognostic indicators

    Br J Cancer

    (1996)
  • V. Pawlowski et al.

    Prognostic value of the type I growth factor receptors in a large series of human primary breast cancers quantified with a real-time reverse transcription-polymerase chain reaction assay

    Clin Cancer Res

    (2000)
  • Y. Lee et al.

    Correlated expression of erbB-3 with hormone receptor expression and favourable clinical outcome in invasive ductal carcinomas of the breast

    Am J Clin Pathol

    (2007)
  • A.K. Koutras et al.

    Evaluation of the prognostic and predictive value of HER family mRNA expression in high-risk early breast cancer

    Br J Cancer

    (2008)
  • H. Lee et al.

    A naturally occurring secreted human ErbB3 receptor isoform inhibits heregulin-stimulated activation of ErbB2, ErbB3, and ErbB4

    Cancer Res

    (2001)
  • Cited by (71)

    • The role of the NDRG1 in the pathogenesis and treatment of breast cancer

      2023, Biochimica et Biophysica Acta - Reviews on Cancer
    • Beyond HER2: Targeting the ErbB receptor family in breast cancer

      2022, Cancer Treatment Reviews
      Citation Excerpt :

      HER3 is encoded by the ERBB3 gene, located on chromosome 12q13; known ligands for this receptor include NRG-1 and NRG-2 [44]. Of special note, HER3 does not possess kinase activity on its own, however it is capable of forming heterodimers with HER2 (and/or EGFR), which dramatically increases transphosphorylation and activation of downstream signaling cascades, in perhaps the most mitogenic stimulus in human breast cancer [44,45]. Moreover HER3 stands out among ErbB family members as a potent inducer of PI3K activity, due to direct binding with the PI3K p85 subunit [46].

    • Development of recombinant biomimetic nano-carrier for targeted gene transfer to HER3 positive breast cancer

      2021, International Journal of Biological Macromolecules
      Citation Excerpt :

      Although this strategy is smart and interesting, it may not be effective in targeting all breast cancer cells. It was reported that the HER3 -mediated signaling is a route for resistance to agents targeting HER2 [41]. Moreover, due to subsequent increases in membrane HER3 expression, the FDA approved HER2 targeted agents are not completely effective [42,43].

    • Negative feedback regulation of ErbB4 tyrosine kinase activity by ERK-mediated non-canonical phosphorylation

      2019, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      ErbB4 has four different isoforms classified by variants at the extracellular juxtamembrane domain (JM-a and JM-b) and the intracellular domain (CYT-1 and CYT-2) (Fig. 1A). Exons encoding JM-a and JM-b are alternatively spliced to produce variants sensitive (JM-a) or resistant (JM-b) to proteolytic cleavage [4,6,32]. Second alternative splicing in the intracellular domain (ICD) results in the insertion of a 16 amino acid CYT region (SEIGHSPPPAYTPMSG) [4,6,11].

    View all citing articles on Scopus

    Angelos K. Koutras, M.D., Ph.D., is a consultant medical oncologist in the Division of Medical Oncology of the University Hospital of Patras, Greece. He is also a senior research fellow at the Laboratory of Clinical Oncology of the University Hospital of Patras, Greece. His main clinical interests include breast, colorectal and lung cancer. His research interests focus on human epidermal growth factor receptor signalling pathways in breast cancer, as well as hormonal signalling in non-small cell lung cancer. Dr. Koutras is a member of the American Society of Clinical Oncology and a certified member of the European Society of Medical Oncology.

    View full text